Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 9(40): 25796-25807, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29899822

ABSTRACT

Alveolar rhabdomyosarcoma (aRMS) is an aggressive subtype of the most common soft tissue cancer in children. A hallmark of aRMS tumors is incomplete myogenic differentiation despite expression of master myogenic regulators such as MyoD. We previously reported that histone methyltransferase KMT1A suppresses MyoD function to maintain an undifferentiated state in aRMS cells, and that loss of KMT1A is sufficient to induce differentiation and suppress malignant phenotypes in these cells. Here, we develop a chemical compound screening approach using MyoD-responsive luciferase reporter myoblast cells to identify compounds that alleviate suppression of MyoD-mediated differentiation by KMT1A. A screen of pharmacological compounds yielded the topoisomerase I (TOP1) poison camptothecin (CPT) as the strongest hit in our assay system. Furthermore, treatment of aRMS cells with clinically relevant CPT derivative irinotecan restores MyoD function, and myogenic differentiation in vitro and in a xenograft model. This differentiated phenotype was associated with downregulation of the KMT1A protein. Remarkably, loss of KMT1A in CPT-treated cells occurs independently of its well-known anti-TOP1 mechanism. We further demonstrate that CPT can directly inhibit KMT1A activity in vitro. Collectively, these findings uncover a novel function of CPT that downregulates KMT1A independently of CPT-mediated TOP1 inhibition and permits differentiation of aRMS cells.

2.
Skelet Muscle ; 6: 28, 2016.
Article in English | MEDLINE | ID: mdl-27551368

ABSTRACT

BACKGROUND: Master transcription factor MyoD can initiate the entire myogenic gene expression program which differentiates proliferating myoblasts into multinucleated myotubes. We previously demonstrated that histone methyltransferase KMT1A associates with and inhibits MyoD in proliferating myoblasts, and must be removed to allow differentiation to proceed. It is known that pro-myogenic signaling pathways such as PI3K/AKT and p38α MAPK play critical roles in enforcing associations between MyoD and transcriptional activators, while removing repressors. However, the mechanism which displaces KMT1A from MyoD, and the signals responsible, remain unknown. METHODS: To investigate the role of p38α on MyoD-mediated differentiation, we utilized C2C12 myoblast cells as an in vitro model. p38α activity was either augmented via overexpression of a constitutively active upstream kinase or blocked via lentiviral delivery of a specific p38α shRNA or treatment with p38α/ß inhibitor SB203580. Overexpression of KMT1A in these cells via lentiviral delivery was also used as a system wherein terminal differentiation is impeded by high levels of KMT1A. RESULTS: The association of KMT1A and MyoD persisted, and differentiation was blocked in C2C12 myoblasts specifically after pharmacologic or genetic blockade of p38α. Conversely, forced activation of p38α was sufficient to activate MyoD and overcome the differentiation blockade in KMT1A-overexpressing C2C12 cells. Consistent with this finding, KMT1A phosphorylation during C2C12 differentiation correlated strongly with the activation of p38α. This phosphorylation was prevented by the inhibition of p38α. Biochemical studies further revealed that KMT1A can be a direct substrate for p38α. Importantly, chromatin immunoprecipitation (ChIP) studies show that the removal of KMT1A-mediated transcription repressive histone tri-methylation (H3K9me3) from the promoter of the Myogenin gene, a critical regulator of muscle differentiation, is dependent on p38α activity in C2C12 cells. Elevated p38α activity was also sufficient to remove this repressive H3K9me3 mark. Moreover, ChIP studies from C2C12 cells show that p38α activity is necessary and sufficient to establish active H3K9 acetylation on the Myogenin promoter. CONCLUSIONS: Activation of p38α displaces KMT1A from MyoD to initiate myogenic gene expression upon induction of myoblasts differentiation.


Subject(s)
Cell Differentiation , Methyltransferases/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , MyoD Protein/metabolism , Myoblasts/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Humans , Mice , Myogenin/genetics , Myogenin/metabolism , Phosphorylation , Promoter Regions, Genetic , Signal Transduction
3.
Mol Cancer Ther ; 12(12): 2663-74, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24107448

ABSTRACT

Alveolar rhabdomyosarcoma comprises a rare highly malignant tumor presumed to be associated with skeletal muscle lineage in children. The hallmark of the majority of alveolar rhabdomyosarcoma is a chromosomal translocation that generates the PAX3-FOXO1 fusion protein, which is an oncogenic transcription factor responsible for the development of the malignant phenotype of this tumor. Alveolar rhabdomyosarcoma cells are dependent on the oncogenic activity of PAX3-FOXO1, and its expression status in alveolar rhabdomyosarcoma tumors correlates with worst patient outcome, suggesting that blocking this activity of PAX3-FOXO1 may be an attractive therapeutic strategy against this fusion-positive disease. In this study, we screened small molecule chemical libraries for inhibitors of PAX3-FOXO1 transcriptional activity using a cell-based readout system. We identified the Sarco/endoplasmic reticulum Ca(2+)-ATPases (SERCA) inhibitor thapsigargin as an effective inhibitor of PAX3-FOXO1. Subsequent experiments in alveolar rhabdomyosarcoma cells showed that activation of AKT by thapsigargin inhibited PAX3-FOXO1 activity via phosphorylation. Moreover, this AKT activation appears to be associated with the effects of thapsigargin on intracellular calcium levels. Furthermore, thapsigargin inhibited the binding of PAX3-FOXO1 to target genes and subsequently promoted its proteasomal degradation. In addition, thapsigargin treatment decreases the growth and invasive capacity of alveolar rhabdomyosarcoma cells while inducing apoptosis in vitro. Finally, thapsigargin can suppress the growth of an alveolar rhabdomyosarcoma xenograft tumor in vivo. These data reveal that thapsigargin-induced activation of AKT is an effective mechanism to inhibit PAX3-FOXO1 and a potential agent for targeted therapy against alveolar rhabdomyosarcoma.


Subject(s)
Oncogene Proteins, Fusion/antagonists & inhibitors , Paired Box Transcription Factors/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Rhabdomyosarcoma, Alveolar/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Phenotype , Phosphorylation/drug effects , Protein Binding , Proteolysis , Small Molecule Libraries , Thapsigargin/pharmacology , Transcription, Genetic , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Cell Cycle ; 11(5): 895-908, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22333587

ABSTRACT

The chimeric PAX3-FKHR transcription factor is present in a majority of alveolar rhabdomyosarcoma (ARMS), an aggressive skeletal muscle cancer of childhood. PAX3-FKHR-mediated aberrant myogenic gene expression resulting in escape from terminal differentiation program is believed to contribute in ARMS development. In skeletal muscle differentiation, activation of AKT pathway leads to myogenic gene activation and terminal differentiation. Here, we report that AKT acts, in part, by modulating PAX3-FKHR transcriptional activity via phosphorylation in the maintenance of the myogenic differentiation blockade in established mouse models of ARMS cells. We observed that low levels of AKT activity are associated with elevated levels of PAX3-FKHR transcriptional activity, and AKT hyperactivation results in PAX3-FKHR phosphorylation coupled with decreased activity once cells are under differentiation-permissible conditions. Subsequent data shows that attenuated AKT activity-associated PAX3-FKHR activity is required to suppress the function of MyoD, a key myogenic regulator of muscle differentiation. Conversely, decreased PAX3-FKHR activity results in the eradication of MyoD expression and subsequent suppression of the myogenic differentiation. Thus, AKT regulation of the PAX3- FKHR suppresses myogenic gene expression in ARMS cells, causing a failure in differentiation. Evidence is presented that provides a novel molecular link between AKT and PAX3-FKHR in maintaining myogenic differentiation blockade in ARMS.


Subject(s)
Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Disease Models, Animal , Gene Knock-In Techniques , Mice , MyoD Protein/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Transcription, Genetic
6.
Cancer Res ; 71(11): 3921-31, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21493592

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) is an aggressive pediatric muscle cancer, which arrested during the process of skeletal muscle differentiation. In muscle myoblast cells, ectopic expression of the histone H3 lysine 9 (H3K9) methytransferase KMT1A blocks differentiation by repressing a myogenic gene expression program. In this study, we tested the hypothesis that activation of a KMT1A-mediated program of transcriptional repression prevents ARMS cells from differentiating. We investigated whether KMT1A represses the expression of differentiation-associated genes in ARMS cells, thereby blocking muscle differentiation. Our results show that expression of KMT1A is induced in human ARMS cancer cell lines when cultured under differentiation-permissible conditions. shRNA-mediated knockdown of KMT1A decreased anchorage dependent and independent cell proliferation and tumor xenograft growth, increased expression of differentiation-associated genes, and promoted the appearance of a terminally differentiated-like phenotype. Finally, shRNA-directed KMT1A knockdown restored the impaired transcriptional activity of the myogenic regulator MyoD. Together, our results suggested that high levels of KMT1A in ARMS cells under differentiation conditions impairs MyoD function, thereby arresting myogenic differentiation in these tumor cells. Thus, targeting KMT1A may be a novel strategy for the treatment of this disease.


Subject(s)
Histone-Lysine N-Methyltransferase/biosynthesis , Rhabdomyosarcoma, Alveolar/enzymology , Rhabdomyosarcoma, Alveolar/pathology , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Child , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , MyoD Protein/metabolism , Myogenin/genetics , Promoter Regions, Genetic , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Rhabdomyosarcoma, Alveolar/genetics , Transduction, Genetic , Transplantation, Heterologous
7.
Oncotarget ; 1(4): 289-303, 2010 Aug.
Article in English | MEDLINE | ID: mdl-21304179

ABSTRACT

Tumorigenic potential of glioblastoma multiforme (GBM) cells is, in part, attributable to their undifferentiated (neural stem cell-like) phenotype. Astrocytic differentiation of GBM cells is associated with transcriptional induction of Glial Fibrillary Acidic Protein (GFAP) and repression of Nestin, whereas the reciprocal transcription program operates in undifferentiated GBM cells. The molecular mechanisms underlying the regulation of these transcription programs remain elusive. Here, we show that the transcriptional co-activator p300 was expressed in GBM tumors and cell lines and acted as an activator of the GFAP gene and a repressor of the Nestin gene. On the other hand, Myc (formerly known as c-Myc overrode these p300 functions by repressing the GFAP gene and inducing the Nestin gene in GBM cells. Moreover, RNAi-mediated inhibition of p300 expression significantly enhanced the invasion potential of GBM cells in vitro. Taken together, these data suggest that dedifferentiated/undifferentiated GBM cells are more invasive than differentiated GBM cells. Because invasion is a major cause of GBM morbidity, differentiation therapy may improve the clinical outcome.


Subject(s)
Cell Differentiation/genetics , E1A-Associated p300 Protein/genetics , Genes, myc , Glioblastoma/genetics , Glioblastoma/pathology , Animals , Blotting, Western , Cell Dedifferentiation , Cell Line, Tumor , Cell Proliferation , E1A-Associated p300 Protein/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glial Fibrillary Acidic Protein/genetics , Glioblastoma/metabolism , Humans , Intermediate Filament Proteins/genetics , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells , Nerve Tissue Proteins/genetics , Nestin , Neural Stem Cells , RNA Interference , Transcription, Genetic
8.
EMBO J ; 25(14): 3323-34, 2006 Jul 26.
Article in English | MEDLINE | ID: mdl-16858404

ABSTRACT

Suv39h1 is a histone H3 lysine-9 (H3-K9) specific methyltransferase (HMT) that is associated with gene silencing through chromatin modification. The transition from proliferation into differentiation of muscle cell is accompanied by transcriptional activation of previously silent muscle genes. I report Suv39h1 interaction with myogenic regulator MyoD in proliferating muscle cells and its HMT activity, which is associated with MyoD, diminishes as differentiation proceeds. The Suv39h1-MyoD complex was detected on the chromatin regulatory regions of a silent differentiation signal muscle gene myogenin and that Suv39h1 presence correlated with H3-K9 methylation. Increased Suv39h1 expression repressed MyoD-dependent muscle gene expression and this property required its HMT activity. This repression required Suv39h1 association with MyoD as well as sustained methylation of H3-K9 on myogenin promoter. Suv39h1 was required for muscle gene repression because its abrogation by siRNA activates these gene expressions by MyoD. These findings suggest that Suv39h1 presence in association with MyoD on the promoter of muscle genes silences gene transcription, providing a necessary checkpoint between proliferation and differentiation.


Subject(s)
Cell Differentiation/physiology , Histone-Lysine N-Methyltransferase/physiology , Methyltransferases/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , MyoD Protein/antagonists & inhibitors , Repressor Proteins/physiology , Animals , Cell Line , Cell Proliferation , HeLa Cells , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Methyltransferases/deficiency , Methyltransferases/genetics , Mice , Mice, Inbred C3H , Mice, Knockout , MyoD Protein/physiology , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...